751
|
Bangash FS, Saeed G, Shahab P, Waheed A. COVID-19: An Update Regarding the Quest for Finding an Effective Cure. Cureus 2020; 12:e9010. [PMID: 32775090 PMCID: PMC7402638 DOI: 10.7759/cureus.9010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/05/2020] [Indexed: 11/23/2022] Open
Abstract
The coronavirus disease (COVID-19) is a global pandemic. COVID-19 is caused by a novel coronavirus known as severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which was identified at the end of 2019 in a cluster of pneumonia cases in Wuhan, China. It has likely affected everyone in the world either directly or indirectly, financially or through social isolation measures. There are now many publications about its etiology, epidemiology, investigations, and clinical presentation. Therefore, the medical community has a much better understanding of the disease as compared to a few months ago. There is no effective, safe treatment for COVID-19. There are many guidelines, clinical trials, and information on various media platforms that hinder the tracking of developments in this rapidly evolving situation. In this review, we provide a detailed update on various emerging treatment options and analyze the results of recent trials. This review also discusses current progress on finding a COVID-19 vaccine.
Collapse
Affiliation(s)
- Fahed S Bangash
- Internal Medicine, Hull University Teaching Hospitals NHS Trust, Hull, GBR
| | - Gulalei Saeed
- Internal Medicine, Shifa International Hospital, Islamabad, PAK
| | | | | |
Collapse
|
752
|
Timis TL, Florian IA, Vesa SC, Orasan RI. Treatment of psoriasis - unburdened by fear during SARS-CoV2 menace. J DERMATOL TREAT 2020; 33:662-665. [PMID: 32594821 DOI: 10.1080/09546634.2020.1788704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In a matter of mere months, humanity was unexpectedly struck by the appearance of SARS-CoV-2, shifting our perception as medical practitioners regarding our day-to-day activity. One especially disconcerting change was patient addressability to medical facilities, as well as access to proper healthcare in various fields. As these changes occurred rapidly, dermatologists too had to adapt by means of teledermatology, giving us back the ability to reach, treat, and comfort our patients. Among the individuals requiring special dermatological attention are those suffering from psoriasis, especially considering that the biological therapies employed in treating this debilitating disease become questionable in the circumstance of the current pandemic. As more evidence surfaces concerning the pathophysiology of SARS-CoV-2, we become closer to understanding which therapies may interfere with its clearance, and which are actually safe to use. This review aims to answer the question, are biological therapies warranted in the treatment of psoriasis during the COVID-19 outbreak, or should they be discontinued?
Collapse
Affiliation(s)
- Teodora-Larisa Timis
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioan-Alexandru Florian
- Department of Neurosciences, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Stefan-Cristian Vesa
- Department of Pharmacology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Remus-Ioan Orasan
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
753
|
Di Micco P, Di Micco G, Russo V, Poggiano MR, Salzano C, Bosevski M, Imparato M, Fontanella L, Fontanella A. Blood Targets of Adjuvant Drugs Against COVID19. J Blood Med 2020; 11:237-241. [PMID: 32694923 PMCID: PMC7338832 DOI: 10.2147/jbm.s256121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
While waiting for the vaccine and/or the best treatment for COVID19, several drugs have been identified as potential adjuvant drugs to counteract the viral action. Several drugs, in fact, have been suggested for their ancillary antiviral role. Viral proteases and peptidases, may interact with well-known drugs such as anticoagulants, antihypertensives, antiserotoninergics and immunomodulants. We here report a basic list of these drugs that include bioflavonoids, heparinoids, ACE inhibitors, angiotensin receptor blockers, antiserotoninergics, and monoclonal antibodies against cytokines that may interact with the viral cycle.
Collapse
Affiliation(s)
- Pierpaolo Di Micco
- Internal Medicine Department, Emergency Room Unit, Fatebenefratelli Hospital of Naples, Naples, Italy
| | | | - Vincenzo Russo
- University Cardiology Clinic, Faculty of Medicine, Skopje, Macedonia
| | - Maria Rita Poggiano
- Internal Medicine Department, Emergency Room Unit, Fatebenefratelli Hospital of Naples, Naples, Italy
| | - Ciro Salzano
- Internal Medicine Department, Emergency Room Unit, Fatebenefratelli Hospital of Naples, Naples, Italy
| | - Marijan Bosevski
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli” Monaldi Hospital, Naples80131, Italy
| | - Michele Imparato
- Internal Medicine Department, Emergency Room Unit, Fatebenefratelli Hospital of Naples, Naples, Italy
| | - Luca Fontanella
- Internal Medicine Department, Emergency Room Unit, Fatebenefratelli Hospital of Naples, Naples, Italy
| | - Andrea Fontanella
- Internal Medicine Department, Emergency Room Unit, Fatebenefratelli Hospital of Naples, Naples, Italy
| |
Collapse
|
754
|
Perricone C, Triggianese P, Bartoloni E, Cafaro G, Bonifacio AF, Bursi R, Perricone R, Gerli R. The anti-viral facet of anti-rheumatic drugs: Lessons from COVID-19. J Autoimmun 2020; 111:102468. [PMID: 32317220 PMCID: PMC7164894 DOI: 10.1016/j.jaut.2020.102468] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023]
Abstract
The outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has posed the world at a pandemic risk. Coronavirus-19 disease (COVID-19) is an infectious disease caused by SARS-CoV-2, which causes pneumonia, requires intensive care unit hospitalization in about 10% of cases and can lead to a fatal outcome. Several efforts are currently made to find a treatment for COVID-19 patients. So far, several anti-viral and immunosuppressive or immunomodulating drugs have demonstrated some efficacy on COVID-19 both in vitro and in animal models as well as in cases series. In COVID-19 patients a pro-inflammatory status with high levels of interleukin (IL)-1B, IL-1 receptor (R)A and tumor necrosis factor (TNF)-α has been demonstrated. Moreover, high levels of IL-6 and TNF-α have been observed in patients requiring intensive-care-unit hospitalization. This provided rationale for the use of anti-rheumatic drugs as potential treatments for this severe viral infection. Other agents, such as hydroxychloroquine and chloroquine might have a direct anti-viral effect. The anti-viral aspect of immunosuppressants towards a variety of viruses has been known since long time and it is herein discussed in the view of searching for a potential treatment for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Elena Bartoloni
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Angelo F Bonifacio
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Roberto Bursi
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Roberto Gerli
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy.
| |
Collapse
|
755
|
Chan AS, Rout A. Use of Neutrophil-to-Lymphocyte and Platelet-to-Lymphocyte Ratios in COVID-19. J Clin Med Res 2020; 12:448-453. [PMID: 32655740 PMCID: PMC7331861 DOI: 10.14740/jocmr4240] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND As the pandemic of coronavirus disease 2019 (COVID-19) continues, prognostic markers are now being identified. The neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are easily accessible values that have been known to correlate with inflammation and prognosis in several conditions. We used the available data to identify the association of NLR and PLR with the severity of COVID-19. METHODS A literature search using EMBASE, MEDLINE, and Google Scholar for studies reporting the use of NLR and PLR in COVID-19 published until April 28, 2020, was performed. Random effects meta-analysis was done to estimate standard mean difference (SMD) of NLR and PLR values with 95% confidence interval (CI) between severe and non-severe COVID-19 cases. RESULTS A total of 20 studies with 3,508 patients were included. Nineteen studies reported NLR values, while five studies reported PLR values between severe and non-severe COVID-19 patients. Higher levels of NLR (SMD: 2.80, 95% CI: 2.12 - 3.48, P < 0.00001) and PLR (SMD: 1.82, 95% CI: 1.03 - 2.61, P < 0.00001)) were seen in patients with severe disease compared to non-severe disease. CONCLUSIONS NLR and PLR can be used as independent prognostic markers of disease severity in COVID-19.
Collapse
Affiliation(s)
- Abigail Sy Chan
- Department of Medicine, Sinai Hospital of Baltimore, Baltimore, MD 21215, USA
| | - Amit Rout
- Department of Medicine, Sinai Hospital of Baltimore, Baltimore, MD 21215, USA
| |
Collapse
|
756
|
Shamdasani P, Trubiano JA, Smibert OC, Owen CE, Liew DFL. COVID
‐19: collaboration will keep us ahead of the curve. Intern Med J 2020; 50:784-786. [PMID: 32656972 PMCID: PMC7404842 DOI: 10.1111/imj.14888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022]
Affiliation(s)
| | - Jason A. Trubiano
- COVID Unit, Department of Infectious DiseasesAustin Health Melbourne Victoria Australia
- Department of MedicineUniversity of Melbourne Melbourne Victoria Australia
- The National Centre for Infections in CancerPeter MacCallum Cancer Centre Melbourne Victoria Australia
| | - Olivia C. Smibert
- COVID Unit, Department of Infectious DiseasesAustin Health Melbourne Victoria Australia
- Department of MedicineUniversity of Melbourne Melbourne Victoria Australia
- The National Centre for Infections in CancerPeter MacCallum Cancer Centre Melbourne Victoria Australia
| | - Claire E. Owen
- Department of RheumatologyAustin Health Melbourne Victoria Australia
- Department of MedicineUniversity of Melbourne Melbourne Victoria Australia
| | - David F. L. Liew
- Department of RheumatologyAustin Health Melbourne Victoria Australia
- Department of MedicineUniversity of Melbourne Melbourne Victoria Australia
- Department of Clinical Pharmacology and TherapeuticsAustin Health Melbourne Victoria Australia
| |
Collapse
|
757
|
Fischer A. Resistance of children to Covid-19. How? Mucosal Immunol 2020; 13:563-565. [PMID: 32467603 PMCID: PMC8851881 DOI: 10.1038/s41385-020-0303-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 02/04/2023]
Affiliation(s)
- Alain Fischer
- Collège de France, Paris, France,Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France,INSERM UMR 1163, Institut Imagine, Paris, France
| |
Collapse
|
758
|
Chen R, Sang L, Jiang M, Yang Z, Jia N, Fu W, Xie J, Guan W, Liang W, Ni Z, Hu Y, Liu L, Shan H, Lei C, Peng Y, Wei L, Liu Y, Hu Y, Peng P, Wang J, Liu J, Chen Z, Li G, Zheng Z, Qiu S, Luo J, Ye C, Zhu S, Zheng J, Zhang N, Li Y, He J, Li J, Li S, Zhong N. Longitudinal hematologic and immunologic variations associated with the progression of COVID-19 patients in China. J Allergy Clin Immunol 2020; 146:89-100. [PMID: 32407836 PMCID: PMC7212968 DOI: 10.1016/j.jaci.2020.05.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Crucial roles of hematologic and immunologic responses in progression of coronavirus disease 2019 (COVID-19) remain largely unclear. OBJECTIVE We sought to address the dynamic changes in hematologic and immunologic biomarkers and their associations with severity and outcomes of COVID-19. METHODS A retrospective study including 548 patients with COVID-19 with clarified outcome (discharged or deceased) from a national cohort in China was performed. Cross-sectional and longitudinal variations were compared and the associations with different severity and outcomes were analyzed. RESULTS On admission, the counts of lymphocytes, T-cell subsets, eosinophils, and platelets decreased markedly, especially in severe/critical and fatal patients. Increased neutrophil count and neutrophils-to-lymphocytes ratio were predominant in severe/critical cases or nonsurvivors. During hospitalization, eosinophils, lymphocytes, and platelets showed an increasing trend in survivors, but maintained lower levels or dropped significantly afterwards in nonsurvivors. Nonsurvivors kept a high level or showed an upward trend for neutrophils, IL-6, procalcitonin, D-dimer, amyloid A protein, and C-reactive protein, which were kept stable or showed a downward trend in survivors. Positive correlation between CD8+ T-cell and lymphocytes count was found in survivors but not in nonsurvivors. A multivariate Cox regression model suggested that restored levels of lymphocytes, eosinophils, and platelets could serve as predictors for recovery, whereas progressive increases in neutrophils, basophils, and IL-6 were associated with fatal outcome. CONCLUSIONS Hematologic and immunologic impairment showed a significantly different profile between survivors and nonsurvivors in patients with COVID-19 with different severity. The longitudinal variations in these biomarkers could serve to predict recovery or fatal outcome.
Collapse
Affiliation(s)
- Ruchong Chen
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Sang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mei Jiang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaowei Yang
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nan Jia
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wanyi Fu
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaxing Xie
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weijie Guan
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhengyi Ni
- Wuhan Jin-yin tan Hospital, Wuhan, Hubei, China
| | - Yu Hu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Liu
- Shenzhen Third People's Hospital, Shenzhen, China
| | - Hong Shan
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Chunliang Lei
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yixiang Peng
- The Central Hospital of Wuhan, Wuhan, Hubei, China
| | - Li Wei
- Wuhan No.1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, Hubei, China
| | - Yong Liu
- Chengdu Public Health Clinical Medical Center, Chengdu, Sichuan, China
| | - Yahua Hu
- Huangshi Central Hospital of Edong Healthcare Group, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
| | - Peng Peng
- Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Jianming Wang
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jiyang Liu
- The First Hospital of Changsha, Changsha, Hunan, China
| | - Zhong Chen
- The Third People's Hospital of Hainan Province, Sanya, Hainan, China
| | - Gang Li
- Huanggang Central Hospital, Huanggang, Hubei, China
| | - Zhijian Zheng
- Wenling First People's Hospital, Wenling, Zhejiang, China
| | - Shaoqin Qiu
- The Third People's Hospital of Yichang, Yichang, Hubei
| | - Jie Luo
- Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | | | - Shaoyong Zhu
- The People's Hospital of Huangpi District, Wuhan, China
| | - Jinping Zheng
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nuofu Zhang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yimin Li
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianxing He
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Li
- Department of Allergy and Clinical Immunology, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shiyue Li
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Nanshan Zhong
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
759
|
Portincasa P, Krawczyk M, Machill A, Lammert F, Di Ciaula A. Hepatic consequences of COVID-19 infection. Lapping or biting? Eur J Intern Med 2020; 77:18-24. [PMID: 32507608 PMCID: PMC7262543 DOI: 10.1016/j.ejim.2020.05.035] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/14/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) starting last December in China placed emphasis on liver involvement during infection. This review discusses the underlying mechanisms linking COVID-19 to liver dysfunction, according to recent available information, while waiting further studies. The manifestations of liver damage are usually mild (moderately elevated serum aspartate aminotransferase activities), and generally asymptomatic. Few patients can still develop severe liver problems, and therapeutic options can be limited. Liver dysfunction may affect about one-third of the patients, with prevalence greater in men than women, and in elderly. Mechanisms of damage are complex and include direct cholangiocyte damage and other coexisting conditions such as the use of antiviral drugs, systemic inflammatory response, respiratory distress syndrome-induced hypoxia, sepsis, and multiple organ dysfunction. During new COVID-19 infections, liver injury may be observed. If liver involvement appears during COVID-19 infection, however, attention is required. This is particularly true if patients are older or have a pre-existing history of liver diseases. During COVID-19 infection, the onset of liver damage impairs the prognosis, and hospital stay is longer.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari 70124, Italy.
| | - Marcin Krawczyk
- Department of Medicine II Saarland University Medical Center, Saarland University, Homburg, Germany; Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Laboratory of Metabolic Liver Diseases, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Antonia Machill
- Department of Medicine II Saarland University Medical Center, Saarland University, Homburg, Germany.
| | - Frank Lammert
- Department of Medicine II Saarland University Medical Center, Saarland University, Homburg, Germany.
| | - Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari 70124, Italy.
| |
Collapse
|
760
|
Konig MF, Powell M, Staedtke V, Bai RY, Thomas DL, Fischer N, Huq S, Khalafallah AM, Koenecke A, Xiong R, Mensh B, Papadopoulos N, Kinzler KW, Vogelstein B, Vogelstein JT, Athey S, Zhou S, Bettegowda C. Preventing cytokine storm syndrome in COVID-19 using α-1 adrenergic receptor antagonists. J Clin Invest 2020; 130:3345-3347. [PMID: 32352407 PMCID: PMC7324164 DOI: 10.1172/jci139642] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Maximilian F. Konig
- Ludwig Center
- Lustgarten Laboratory, and
- Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
- Division of Rheumatology
- Johns Hopkins University (JHU) School of Medicine, Baltimore, Maryland, USA
| | - Mike Powell
- Department of Biomedical Engineering, Institute of Computational Medicine, JHU, Baltimore, Maryland, USA
| | | | | | - David L. Thomas
- Division of Infectious Diseases, JHU School of Medicine, Baltimore, Maryland, USA
| | - Nicole Fischer
- Johns Hopkins University (JHU) School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Ruoxuan Xiong
- Management Science and Engineering, Stanford University, Stanford, California, USA
| | - Brett Mensh
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Nickolas Papadopoulos
- Ludwig Center
- Lustgarten Laboratory, and
- Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Kenneth W. Kinzler
- Ludwig Center
- Lustgarten Laboratory, and
- Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Bert Vogelstein
- Ludwig Center
- Lustgarten Laboratory, and
- Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Joshua T. Vogelstein
- Department of Biomedical Engineering, Institute of Computational Medicine, JHU, Baltimore, Maryland, USA
| | - Susan Athey
- Stanford Graduate School of Business, Stanford University, Stanford, California, USA
| | - Shibin Zhou
- Ludwig Center
- Lustgarten Laboratory, and
- Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Chetan Bettegowda
- Ludwig Center
- Lustgarten Laboratory, and
- Howard Hughes Medical Institute, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
- Department of Neurosurgery, and
| |
Collapse
|
761
|
Solerte SB, Di Sabatino A, Galli M, Fiorina P. Dipeptidyl peptidase-4 (DPP4) inhibition in COVID-19. Acta Diabetol 2020; 57:779-783. [PMID: 32506195 PMCID: PMC7275134 DOI: 10.1007/s00592-020-01539-z] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 02/08/2023]
Abstract
AIMS SARS-CoV-2 causes severe respiratory syndrome (COVID-19) with high mortality due to a direct cytotoxic viral effect and a severe systemic inflammation. We are herein discussing a possible novel therapeutic tool for COVID-19. METHODS Virus binds to the cell surface receptor ACE2; indeed, recent evidences suggested that SARS-CoV-2 may be using as co-receptor, when entering the cells, the same one used by MERS-Co-V, namely the DPP4/CD26 receptor. The aforementioned observation underlined that mechanism of cell entry is supposedly similar among different coronavirus, that the co-expression of ACE2 and DPP4/CD26 could identify those cells targeted by different human coronaviruses and that clinical complications may be similar. RESULTS The DPP4 family/system was implicated in various physiological processes and diseases of the immune system, and DPP4/CD26 is variously expressed on epithelia and endothelia of the systemic vasculature, lung, kidney, small intestine and heart. In particular, DPP4 distribution in the human respiratory tract may facilitate the entrance of the virus into the airway tract itself and could contribute to the development of cytokine storm and immunopathology in causing fatal COVID-19 pneumonia. CONCLUSIONS The use of DPP4 inhibitors, such as gliptins, in patients with COVID-19 with, or even without, type 2 diabetes, may offer a simple way to reduce the virus entry and replication into the airways and to hamper the sustained cytokine storm and inflammation within the lung in patients diagnosed with COVID-19 infection.
Collapse
Affiliation(s)
- Sebastiano Bruno Solerte
- Geriatric and Diabetology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- Internal Medicine Unit, University of Pavia and IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimo Galli
- Department of Biomedical, Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università Degli Studi di Milano, Milan, Italy.
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy.
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
762
|
Baba H, Kanamori H, Oshima K, Seike I, Niitsuma-Sugaya I, Takei K, Sato Y, Tokuda K, Aoyagi T. Prolonged presence of SARS-CoV-2 in a COVID-19 case with rheumatoid arthritis taking iguratimod treated with ciclesonide. J Infect Chemother 2020; 26:1100-1103. [PMID: 32631736 PMCID: PMC7328613 DOI: 10.1016/j.jiac.2020.06.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
We report a coronavirus disease 2019 (COVID-19) case with rheumatoid arthritis taking iguratimod. The patient who continued iguratimod therapy without dose reduction was treated with ciclesonide had an uneventful clinical course, but prolonged detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was observed after resolution of symptoms. The effects of disease-modifying antirheumatic drugs (DMARDs) and ciclesonide on clinical course and viral shedding remain unknown and warrant further investigation.
Collapse
Affiliation(s)
- Hiroaki Baba
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Hajime Kanamori
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan; Division of Infection Control, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Kengo Oshima
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Issei Seike
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Ikumi Niitsuma-Sugaya
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Kentaro Takei
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Yukio Sato
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan; Sendai Medical Imaging Clinic, 2-1-25 Itsutsubashi, Aoba-ku, Sendai, Miyagi, 980-0022, Japan
| | - Koichi Tokuda
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan; Division of Infection Control, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Tetsuji Aoyagi
- Department of Infectious Diseases, Internal Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
763
|
Abstract
The current coronavirus pandemic is an ongoing global health crisis due to COVID-19, caused by severe acute respiratory syndrome coronavirus 2. Although COVID-19 leads to little or mild flu-like symptoms in the majority of affected patients, the disease may cause severe, frequently lethal complications such as progressive pneumonia, acute respiratory distress syndrome and organ failure driven by hyperinflammation and a cytokine storm syndrome. This situation causes various major challenges for gastroenterology. In the context of IBD, several key questions arise. For instance, it is an important question to understand whether patients with IBD (eg, due to intestinal ACE2 expression) might be particularly susceptible to COVID-19 and the cytokine release syndrome associated with lung injury and fatal outcomes. Another highly relevant question is how to deal with immunosuppression and immunomodulation during the current pandemic in patients with IBD and whether immunosuppression affects the progress of COVID-19. Here, the current understanding of the pathophysiology of COVID-19 is reviewed with special reference to immune cell activation. Moreover, the potential implications of these new insights for immunomodulation and biological therapy in IBD are discussed.
Collapse
Affiliation(s)
- Markus F Neurath
- First Department of Medicine and Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91052, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
764
|
So H, Mok CC. COVID-19 and Rheumatic Diseases: Practical Issues. JOURNAL OF CLINICAL RHEUMATOLOGY AND IMMUNOLOGY 2020. [DOI: 10.1142/s2661341720300025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
On March 12, 2020, the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) a pandemic. The rapidly increasing number of cases and deaths have overwhelmed the health care system worldwide. We aimed to provide a narrative review on some practical issues of COVID-19 and rheumatic diseases with the limited data to the date of April 26, 2020.
Collapse
Affiliation(s)
- Ho So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, China
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
| |
Collapse
|
765
|
Di Ciaula A, Palmieri VO, Migliore G, Portincasa P. COVID-19, internists and resilience: the north-south Italy outbreak. Eur J Clin Invest 2020; 50:e13299. [PMID: 32480424 PMCID: PMC7300465 DOI: 10.1111/eci.13299] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Agostino Di Ciaula
- Department of Biomedical Sciences and Human OncologyClinica Medica “A. Murri”University of Bari “Aldo Moro”BariItaly
- International Society of Doctors for Environment (ISDE)ArezzoItaly
| | - Vincenzo O. Palmieri
- Department of Biomedical Sciences and Human OncologyClinica Medica “A. Murri”University of Bari “Aldo Moro”BariItaly
| | - Giovanni Migliore
- General DirectoratePoliclinico‐Giovanni XXIII Regional HospitalBariItaly
| | - Piero Portincasa
- Department of Biomedical Sciences and Human OncologyClinica Medica “A. Murri”University of Bari “Aldo Moro”BariItaly
| | | |
Collapse
|
766
|
Giammaria D, Pajewski A. Can early treatment of patients with risk factors contribute to managing the COVID-19 pandemic? J Glob Health 2020; 10:010377. [PMID: 32582439 PMCID: PMC7307801 DOI: 10.7189/jogh.10.010377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Daniele Giammaria
- Ophthalmology Department, Azienda "Ospedali Riuniti Marche Nord", Pesaro, Italy
| | | |
Collapse
|
767
|
Koralnik IJ, Tyler KL. COVID-19: A Global Threat to the Nervous System. Ann Neurol 2020; 88:1-11. [PMID: 32506549 PMCID: PMC7300753 DOI: 10.1002/ana.25807] [Citation(s) in RCA: 310] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022]
Abstract
In less than 6 months, the severe acute respiratory syndrome-coronavirus type 2 (SARS-CoV-2) has spread worldwide infecting nearly 6 million people and killing over 350,000. Initially thought to be restricted to the respiratory system, we now understand that coronavirus disease 2019 (COVID-19) also involves multiple other organs, including the central and peripheral nervous system. The number of recognized neurologic manifestations of SARS-CoV-2 infection is rapidly accumulating. These may result from a variety of mechanisms, including virus-induced hyperinflammatory and hypercoagulable states, direct virus infection of the central nervous system (CNS), and postinfectious immune mediated processes. Example of COVID-19 CNS disease include encephalopathy, encephalitis, acute disseminated encephalomyelitis, meningitis, ischemic and hemorrhagic stroke, venous sinus thrombosis, and endothelialitis. In the peripheral nervous system, COVID-19 is associated with dysfunction of smell and taste, muscle injury, the Guillain-Barre syndrome, and its variants. Due to its worldwide distribution and multifactorial pathogenic mechanisms, COVID-19 poses a global threat to the entire nervous system. Although our understanding of SARS-CoV-2 neuropathogenesis is still incomplete and our knowledge is evolving rapidly, we hope that this review will provide a useful framework and help neurologists in understanding the many neurologic facets of COVID-19. ANN NEUROL 2020;88:1-11 ANN NEUROL 2020;88:1-11.
Collapse
MESH Headings
- Betacoronavirus
- Brain Diseases/etiology
- Brain Diseases/physiopathology
- Brain Ischemia/etiology
- Brain Ischemia/physiopathology
- COVID-19
- Coronavirus Infections/complications
- Coronavirus Infections/physiopathology
- Disseminated Intravascular Coagulation/etiology
- Disseminated Intravascular Coagulation/physiopathology
- Encephalitis/etiology
- Encephalitis/physiopathology
- Encephalomyelitis, Acute Disseminated/etiology
- Encephalomyelitis, Acute Disseminated/physiopathology
- Guillain-Barre Syndrome/etiology
- Guillain-Barre Syndrome/physiopathology
- Humans
- Inflammation
- Intracranial Hemorrhages/etiology
- Intracranial Hemorrhages/physiopathology
- Leukoencephalitis, Acute Hemorrhagic/etiology
- Leukoencephalitis, Acute Hemorrhagic/physiopathology
- Meningitis, Viral/etiology
- Meningitis, Viral/physiopathology
- Nervous System Diseases/etiology
- Nervous System Diseases/physiopathology
- Olfaction Disorders/etiology
- Olfaction Disorders/physiopathology
- Pandemics
- Pneumonia, Viral/complications
- Pneumonia, Viral/physiopathology
- SARS-CoV-2
- Sinus Thrombosis, Intracranial/etiology
- Sinus Thrombosis, Intracranial/physiopathology
- Stroke/etiology
- Stroke/physiopathology
- Thrombophilia/etiology
- Thrombophilia/physiopathology
Collapse
Affiliation(s)
- Igor J. Koralnik
- Davee Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Kenneth L. Tyler
- Department of Neurology, Medicine, and Immunology‐MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| |
Collapse
|
768
|
Mello MTD, Silva A, Guerreiro RDC, da-Silva FR, Esteves AM, Poyares D, Piovezan R, Treptow E, Starling M, Rosa DS, Pires GN, Andersen ML, Tufik S. Sleep and COVID-19: considerations about immunity, pathophysiology, and treatment. Sleep Sci 2020; 13:199-209. [PMID: 33381288 PMCID: PMC7755266 DOI: 10.5935/1984-0063.20200062] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022] Open
Abstract
The fear and uncertainty caused by the coronavirus disease 2019 (COVID-19) pandemic, threats to survival are one of the main problems of everyday life; however, mental health care must also be considered a priority. During social isolation also called self-quarantine, the restricted mobility and social contact, concern about financial resources and availability of supplies, fear of infection, questions about the duration of self-quarantine, cause anxiety, depression, stress, insomnia and reduced the quality and quantity of sleep, that may present a greater risk to the health of the general population. Sleep disorders are increasingly becoming a major health issue in modern society, and are influenced by retinal stimulation by electronic devices, as well extended and/or night shift-work, which may aggravate the systemic and lung inflammation during viral infections. Sleep disorders can induce pro-inflammatory states and be harmful during the COVID-19 pandemic. The possible interactions between many drugs used to treat COVID-19, and those used to treat sleep disorders are unknown, mostly due to the lack of a standard protocol to treat these patients. Insufficient sleep or irregular sleep-wake cycles may impair health, immune system, induce pro-inflammation state, and may lead to increased vulnerability to viral infections, involving inflammatory and oxidative/antioxidant imbalance. In this sense, obstructive sleep apnea has been associated with recognized COVID-19 risk comorbidities and considered a risk factor for COVID-19. During the COVID-19 pandemic, health care cannot stop, and telemedicine has presented itself as an alternative method of delivering services. When a face-to-face visit is mandatory, or in locations with minimal community transmission where sleep centers have resumed activities, it is important that the sleep center facilities are properly prepared to receive the patients during the COVID-19 pandemic, and follow all relevant safety rules. In this work we gathered a group of researchers, specialists in aspects related to chronobiology, sleep, sleep disorders, and the immune system. Thus, we conducted a narrative review in order to address the relationship between COVID-19 and sleep, as well as its immunological aspects and strategies that may be applied in order to mitigate the harmful effects on health that affects everyone during the pandemic.
Collapse
Affiliation(s)
- Marco Túlio De Mello
- Universidade Federal de Minas Gerais, Departamento de Esportes - Belo Horizonte - Minas Gerais - Brazil
| | - Andressa Silva
- Universidade Federal de Minas Gerais, Departamento de Esportes - Belo Horizonte - Minas Gerais - Brazil
| | | | - Flavia Rodrigues da-Silva
- Universidade Federal de Minas Gerais, Departamento de Esportes - Belo Horizonte - Minas Gerais - Brazil
| | - Andrea Maculano Esteves
- Universidade Estadual de Campinas, Faculdade de Ciências Aplicadas - Limeira - São Paulo - Brazil
| | - Dalva Poyares
- Universidade Federal de São Paulo, Departamento de Psicobiologia - São Paulo - São Paulo - Brazil
| | - Ronaldo Piovezan
- Universidade Federal de São Paulo, Departamento de Psicobiologia - São Paulo - São Paulo - Brazil
| | - Erika Treptow
- Universidade Federal de São Paulo, Departamento de Psicobiologia - São Paulo - São Paulo - Brazil
| | - Marcelo Starling
- Universidade Federal de Minas Gerais, Departamento de Esportes - Belo Horizonte - Minas Gerais - Brazil
| | - Daniela Santoro Rosa
- Federal University of São Paulo, Departament of Microbiology, Imunology and Parasitology - São Paulo - São Paulo - Brazil
| | - Gabriel Natan Pires
- Universidade Federal de São Paulo, Departamento de Psicobiologia - São Paulo - São Paulo - Brazil
- Santa Casa de São Paulo School of Medical Sciences, Department of Physiological Sciences - São Paulo - São Paulo - Brazil
| | - Monica Levy Andersen
- Universidade Federal de São Paulo, Departamento de Psicobiologia - São Paulo - São Paulo - Brazil
| | - Sergio Tufik
- Universidade Federal de São Paulo, Departamento de Psicobiologia - São Paulo - São Paulo - Brazil
| |
Collapse
|
769
|
Manjili RH, Zarei M, Habibi M, Manjili MH. COVID-19 as an Acute Inflammatory Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:12-19. [PMID: 32423917 PMCID: PMC7333792 DOI: 10.4049/jimmunol.2000413] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023]
Abstract
The 2019 coronavirus disease (COVID-19) pandemic caused by the virus severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has created an unprecedented global crisis for the infrastructure sectors, including economic, political, healthcare, education, and research systems. Although over 90% of infected individuals are asymptomatic or manifest noncritical symptoms and will recover from the infection, those individuals presenting with critical symptoms are in urgent need of effective treatment options. Emerging data related to mechanism of severity and potential therapies for patients presenting with severe symptoms are scattered and therefore require a comprehensive analysis to focus research on developing effective therapeutics. A critical literature review suggests that the severity of SARS-CoV-2 infection is associated with dysregulation of inflammatory immune responses, which in turn inhibits the development of protective immunity to the infection. Therefore, the use of therapeutics that modulate inflammation without compromising the adaptive immune response could be the most effective therapeutic strategy.
Collapse
Affiliation(s)
| | - Melika Zarei
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| | - Mehran Habibi
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 20215
| | - Masoud H Manjili
- Department of Microbiology and Immunology, VCU Institute of Molecular Medicine, VCU School of Medicine, Richmond, VA 23298; and
- VCU Massey Cancer Center, Richmond, VA 23298
| |
Collapse
|
770
|
Radbel J, Narayanan N, Bhatt PJ. Use of Tocilizumab for COVID-19-Induced Cytokine Release Syndrome: A Cautionary Case Report. Chest 2020; 158:e15-e19. [PMID: 32343968 PMCID: PMC7195070 DOI: 10.1016/j.chest.2020.04.024] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
Novel coronavirus disease 2019 (COVID-19) emerged in late December 2019 in Wuhan, China. Since then, COVID-19 has become a pandemic affecting more than 4.1 million people worldwide. Patients with COVID-19 have a wide spectrum of manifestations, one being cytokine release syndrome (CRS) and its fatal correlate, secondary hemophagocytic lymphohistiocytosis (sHLH). Anti-cytokine therapy such as tocilizumab, an IL-6 receptor antagonist, is a potential treatment for COVID-19; however, data regarding the efficacy of this anti-IL-6 therapy are currently lacking. We report two cases of patients who received a diagnosis of COVID-19 complicated by CRS and were treated with tocilizumab. Both patients progressed to sHLH despite treatment with tocilizumab, and one developed viral myocarditis, challenging the safety and clinical usefulness of tocilizumab in the treatment of COVID-19-induced CRS. These cases highlight the need for clinical trials to determine optimal patient selection and timing for the use of tocilizumab during this disease process.
Collapse
MESH Headings
- Adult
- Aged
- Anti-Infective Agents/administration & dosage
- Anti-Inflammatory Agents/administration & dosage
- Antibodies, Monoclonal, Humanized/administration & dosage
- Azithromycin/administration & dosage
- Betacoronavirus/isolation & purification
- C-Reactive Protein/analysis
- COVID-19
- Clinical Deterioration
- Coronavirus Infections/complications
- Coronavirus Infections/diagnosis
- Coronavirus Infections/physiopathology
- Coronavirus Infections/therapy
- Cytokine Release Syndrome/blood
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/virology
- Fatal Outcome
- Female
- Humans
- Hydroxychloroquine/administration & dosage
- Hypoxia/etiology
- Hypoxia/therapy
- Lymphohistiocytosis, Hemophagocytic/blood
- Lymphohistiocytosis, Hemophagocytic/therapy
- Lymphohistiocytosis, Hemophagocytic/virology
- Male
- Myocarditis/therapy
- Myocarditis/virology
- Pandemics
- Pneumonia, Viral/complications
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/etiology
- Pneumonia, Viral/physiopathology
- Pneumonia, Viral/therapy
- Respiration, Artificial/methods
- SARS-CoV-2
- Shock, Septic/etiology
- Shock, Septic/therapy
Collapse
Affiliation(s)
- Jared Radbel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ.
| | - Navaneeth Narayanan
- Department of Pharmacy Practice and Administration, Rutgers-Ernest Mario School of Pharmacy, New Brunswick, NJ
| | - Pinki J Bhatt
- Division of Infectious Disease, Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
771
|
Carter-Timofte ME, Jørgensen SE, Freytag MR, Thomsen MM, Brinck Andersen NS, Al-Mousawi A, Hait AS, Mogensen TH. Deciphering the Role of Host Genetics in Susceptibility to Severe COVID-19. Front Immunol 2020; 11:1606. [PMID: 32695122 PMCID: PMC7338588 DOI: 10.3389/fimmu.2020.01606] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/16/2020] [Indexed: 01/19/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) describes a set of symptoms that develop following infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Whilst COVID-19 disease is most serious in patients with significant co-morbidities, the reason for healthy individuals succumbing to fulminant infection is largely unexplained. In this review, we discuss the most recent findings in terms of clinical features and the host immune response, and suggest candidate immune pathways that may be compromised in otherwise healthy individuals with fulminating COVID-19. On the basis of this early knowledge we reason a potential genetic effect on host immune response pathways leading to increased susceptibility to SARS-CoV-2 infection. Understanding these pathways may help not only in unraveling disease pathogenesis, but also in suggesting targets for therapy and prophylaxis. Importantly such insight should instruct efforts to identify those at increased risk in order to institute preventative measures, such as prophylactic medication and/or vaccination, when such opportunities arise in the later phases of the current pandemic or during future similar pandemics.
Collapse
Affiliation(s)
- Madalina Elena Carter-Timofte
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Sofie Eg Jørgensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Mette Ratzer Freytag
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Michelle Mølgaard Thomsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Nanna-Sophie Brinck Andersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Ali Al-Mousawi
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Alon Schneider Hait
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Trine H. Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Clinical Medicine, Aarhus, Denmark
| |
Collapse
|
772
|
Kolilekas L, Loverdos K, Giannakaki S, Vlassi L, Levounets A, Zervas E, Gaga M. Can steroids reverse the severe COVID-19 induced "cytokine storm"? J Med Virol 2020; 92:2866-2869. [PMID: 32530507 PMCID: PMC7307112 DOI: 10.1002/jmv.26165] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 01/07/2023]
Abstract
Severe coronavirus disease (COVID-19) is characterized by an excessive proinflammatory cytokine storm, resulting in acute lung injury and development of acute respiratory distress syndrome (ARDS). The role of corticosteroids is controversial in severe COVID-19 pneumonia and associated hyper-inflammatory syndrome. We reported a case series of six consecutive COVID-19 patients with severe pneumonia, ARDS and laboratory indices of hyper-inflammatory syndrome. All patients were treated early with a short course of corticosteroids, and clinical outcomes were compared before and after corticosteroids administration. All patients evaded intubation and intensive care admission, ARDS resolved within 11.8 days (median), viral clearance was achieved in four patients within 17.2 days (median), and all patients were discharged from the hospital in 16.8 days (median). Early administration of short course corticosteroids improves clinical outcome of patients with severe COVID-19 pneumonia and evidence of immune hyperreactivity.
Collapse
Affiliation(s)
- Lykourgos Kolilekas
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| | - Konstantinos Loverdos
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| | - Styliani Giannakaki
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| | - Lamprini Vlassi
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| | - Anastasia Levounets
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| | - Eleftherios Zervas
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| | - Mina Gaga
- 7th Respiratory Medicine Department and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece
| |
Collapse
|
773
|
Malik YS, Kumar N, Sircar S, Kaushik R, Bhat S, Dhama K, Gupta P, Goyal K, Singh MP, Ghoshal U, El Zowalaty ME, R. VO, Yatoo MI, Tiwari R, Pathak M, Patel SK, Sah R, Rodriguez-Morales AJ, Ganesh B, Kumar P, Singh RK. Coronavirus Disease Pandemic (COVID-19): Challenges and a Global Perspective. Pathogens 2020; 9:E519. [PMID: 32605194 PMCID: PMC7400054 DOI: 10.3390/pathogens9070519] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023] Open
Abstract
The technology-driven world of the 21st century is currently confronted with a major threat to humankind, represented by the coronavirus disease (COVID-19) pandemic, caused by the severe acute respiratory syndrome, coronavirus-2 (SARS-CoV-2). As of now, COVID-19 has affected more than 6 million confirmed cases and took 0.39 million human lives. SARS-CoV-2 spreads much faster than its two ancestors, SARS-CoV and Middle East respiratory syndrome-CoV (MERS-CoV), but has low fatality rates. Our analyses speculate that the efficient replication and transmission of SARS-CoV-2 might be due to the high-density basic amino acid residues, preferably positioned in close proximity at both the furin-like cleavage sites (S1/S2 and S2') within the spike protein. Given the high genomic similarities of SARS-CoV-2 to bat SARS-like CoVs, it is likely that bats serve as a reservoir host for its progenitor. Women and children are less susceptible to SARS-CoV-2 infection, while the elderly and people with comorbidities are more prone to serious clinical outcomes, which may be associated with acute respiratory distress syndrome (ARDS) and cytokine storm. The cohesive approach amongst researchers across the globe has delivered high-end viral diagnostics. However, home-based point-of-care diagnostics are still under development, which may prove transformative in current COVID-19 pandemic containment. Similarly, vaccines and therapeutics against COVID-19 are currently in the pipeline for clinical trials. In this review, we discuss the noteworthy advancements, focusing on the etiological viral agent, comparative genomic analysis, population susceptibility, disease epidemiology and diagnosis, animal reservoirs, laboratory animal models, disease transmission, therapeutics, vaccine challenges, and disease mitigation measures.
Collapse
Affiliation(s)
- Yashpal Singh Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; (S.S.); (S.B.)
| | - Naveen Kumar
- ICAR-National Institute of High Security Animal Diseases, OIE Reference Laboratory for Avian Influenza, Bhopal, Madhya Pradesh 462 022, India;
| | - Shubhankar Sircar
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; (S.S.); (S.B.)
| | - Rahul Kaushik
- Laboratory for Structural Bioinformatics, Biosystems Dynamics Research Center, Riken 250-0047, Japan;
| | - Sudipta Bhat
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; (S.S.); (S.B.)
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; (M.P.); (S.K.P.)
| | - Parakriti Gupta
- Medical Microbiology, Department of Virology, PGIMER, Chandigarh 160012, India; (P.G.); (K.G.); (M.P.S.)
| | - Kapil Goyal
- Medical Microbiology, Department of Virology, PGIMER, Chandigarh 160012, India; (P.G.); (K.G.); (M.P.S.)
| | - Mini P. Singh
- Medical Microbiology, Department of Virology, PGIMER, Chandigarh 160012, India; (P.G.); (K.G.); (M.P.S.)
| | - Ujjala Ghoshal
- Department of Microbiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, Uttar Pradesh 226014, India;
| | - Mohamed E. El Zowalaty
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-75 123 Uppsala, Sweden;
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, UAE
| | - VinodhKumar O. R.
- Division of Epidemiology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Mohd Iqbal Yatoo
- Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Srinagar, Jammu and Kashmir 190025, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura, Uttar Pradesh 281001, India;
| | - Mamta Pathak
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; (M.P.); (S.K.P.)
| | - Shailesh Kumar Patel
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India; (M.P.); (S.K.P.)
| | - Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu P.O. BOX 1524, Nepal;
| | - Alfonso J. Rodriguez-Morales
- Public Health and Infection Research Group, Faculty of Health Sciences, Universidad Tecnologica de Pereira, Pereira 660001, Colombia;
- Grupo de Investigacion Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de las Americas, Pereira, Risaralda 660003, Colombia
| | - Balasubramanian Ganesh
- Laboratory Division, Indian Council of Medical Research -National Institute of Epidemiology, Ministry of Health & Family Welfare, Ayapakkam, Chennai, Tamil Nadu 600077, India;
| | - Prashant Kumar
- Amity Institute of Virology and Immunology, J-3 Block, Amity University, Sector-125, Noida, Uttar Pradesh 201303, India;
| | - Raj Kumar Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India;
| |
Collapse
|
774
|
Shen Q, Xiao X, Aierken A, Yue W, Wu X, Liao M, Hua J. The ACE2 expression in Sertoli cells and germ cells may cause male reproductive disorder after SARS-CoV-2 infection. J Cell Mol Med 2020; 24:9472-9477. [PMID: 32594644 PMCID: PMC7361928 DOI: 10.1111/jcmm.15541] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022] Open
Abstract
The serious coronavirus disease‐2019 (COVID‐19) was first reported in December 2019 in Wuhan, China. COVID‐19 is an infectious disease caused by severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2). Angiotensin converting enzyme 2(ACE2) is the cellular receptor for SARS‐CoV‐2. Considering the critical roles of testicular cells for the transmission of genetic information between generations, we analyzed single‐cell RNA‐sequencing (scRNA‐seq) data of adult human testis. The mRNA expression of ACE2 was expressed in both germ cells and somatic cells. Moreover, the positive rate of ACE2 in testes of infertile men was higher than normal, which indicates that SARS‐CoV‐2 may cause reproductive disorders through pathway activated by ACE2 and the men with reproductive disorder may easily to be infected by SARS‐CoV‐2. The expression level of ACE2 was related to the age, and the mid‐aged with higher positive rate than young men testicular cells. Taken together, this research provides a biological background of the potential route for infection of SARS‐CoV‐2 and may enable rapid deciphering male‐related reproductive disorders induced by COVID‐19.
Collapse
Affiliation(s)
- Qiaoyan Shen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Xia Xiao
- College of LifeSciences, Northwest A&F University, Yangling, China
| | - Aili Aierken
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Wei Yue
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Xiaojie Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Mingzhi Liao
- College of LifeSciences, Northwest A&F University, Yangling, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
775
|
Shaikh F, Andersen MB, Sohail MR, Mulero F, Awan O, Dupont-Roettger D, Kubassova O, Dehmeshki J, Bisdas S. Current Landscape of Imaging and the Potential Role for Artificial Intelligence in the Management of COVID-19. Curr Probl Diagn Radiol 2020; 50:430-435. [PMID: 32703538 PMCID: PMC7320858 DOI: 10.1067/j.cpradiol.2020.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023]
Abstract
The clinical management of COVID-19 is challenging. Medical imaging plays a critical role in the early detection, clinical monitoring and outcomes assessment of this disease. Chest x-ray radiography and computed tomography) are the standard imaging modalities used for the structural assessment of the disease status, while functional imaging (namely, positron emission tomography) has had limited application. Artificial intelligence can enhance the predictive power and utilization of these imaging approaches and new approaches focusing on detection, stratification and prognostication are showing encouraging results. We review the current landscape of these imaging modalities and artificial intelligence approaches as applied in COVID-19 management.
Collapse
Affiliation(s)
- Faiq Shaikh
- Image Analysis Group, Philadelphia, PA, USA.
| | - Michael Brun Andersen
- Aarhus University, Aarhus, Denmark; Herlev Gentofte Hospital, The Capital Region, Denmark
| | | | | | - Omer Awan
- University of Maryland, Baltimore, MD
| | | | | | - Jamshid Dehmeshki
- Image Analysis Group, Philadelphia, PA, USA; Kingston University, Kingston-upon-Thames, UK
| | | |
Collapse
|
776
|
Pearce L, Davidson SM, Yellon DM. The cytokine storm of COVID-19: a spotlight on prevention and protection. Expert Opin Ther Targets 2020; 24:723-730. [PMID: 32594778 DOI: 10.1080/14728222.2020.1783243] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The cytokine release syndrome (CRS) of COVID-19 is associated with the development of critical illness requiring multi-organ support. Further research is required to halt progression of multi-organ injury induced by hyper-inflammation. AREAS COVERED PubMed/MEDLINETM databases were accessed between May 9th-June 9th, 2020, to review the latest perspectives on the treatment and pathogenesis of CRS. EXPERT OPINION Over-activity of chemotaxis triggers a macrophage activation syndrome (MAS) resulting in the release of pro-inflammatory cytokines. IL-6 and TNF- α are at the forefront of hyper-inflammation. The inflammatory cascade induces endothelial activation and capillary leak, leading to circulatory collapse and shock. As endothelial dysfunction persists, there is activation of the clotting cascade and microvascular obstruction. Continued endothelial activation results in multi-organ failure, regardless of pulmonary tissue damage. We propose that targeting the endothelium may interrupt this cycle. Immuno-modulating therapies have been suggested, however, further data is necessary to confirm that they do not jeopardize adaptive immunity. Inhibition of IL-6 and the Janus Kinase, signal transducer and activator of transcription proteins pathway (JAK/STAT), are favorable targets. Remote ischemic conditioning (RIC) reduces the inflammation of sepsis in animal models and should be considered as a low risk intervention, in combination with cardiovascular protection.
Collapse
Affiliation(s)
- Lucie Pearce
- The Hatter Cardiovascular Institute, University College London , London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London , London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London , London, UK
| |
Collapse
|
777
|
Tarasova O, Ivanov S, Filimonov DA, Poroikov V. Data and Text Mining Help Identify Key Proteins Involved in the Molecular Mechanisms Shared by SARS-CoV-2 and HIV-1. Molecules 2020; 25:E2944. [PMID: 32604797 PMCID: PMC7357070 DOI: 10.3390/molecules25122944] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
Viruses can be spread from one person to another; therefore, they may cause disorders in many people, sometimes leading to epidemics and even pandemics. New, previously unstudied viruses and some specific mutant or recombinant variants of known viruses constantly appear. An example is a variant of coronaviruses (CoV) causing severe acute respiratory syndrome (SARS), named SARS-CoV-2. Some antiviral drugs, such as remdesivir as well as antiretroviral drugs including darunavir, lopinavir, and ritonavir are suggested to be effective in treating disorders caused by SARS-CoV-2. There are data on the utilization of antiretroviral drugs against SARS-CoV-2. Since there are many studies aimed at the identification of the molecular mechanisms of human immunodeficiency virus type 1 (HIV-1) infection and the development of novel therapeutic approaches against HIV-1, we used HIV-1 for our case study to identify possible molecular pathways shared by SARS-CoV-2 and HIV-1. We applied a text and data mining workflow and identified a list of 46 targets, which can be essential for the development of infections caused by SARS-CoV-2 and HIV-1. We show that SARS-CoV-2 and HIV-1 share some molecular pathways involved in inflammation, immune response, cell cycle regulation.
Collapse
Affiliation(s)
- Olga Tarasova
- Department for Bioinformatics, Institute of Biomedical Chemistry, 107076 Moscow, Russia; (S.I.); (D.A.F.); (V.P.)
| | - Sergey Ivanov
- Department for Bioinformatics, Institute of Biomedical Chemistry, 107076 Moscow, Russia; (S.I.); (D.A.F.); (V.P.)
- Department of Bioinformatics of Pirogov Russian National Research Medical University, 107076 Moscow, Russia
| | - Dmitry A. Filimonov
- Department for Bioinformatics, Institute of Biomedical Chemistry, 107076 Moscow, Russia; (S.I.); (D.A.F.); (V.P.)
| | - Vladimir Poroikov
- Department for Bioinformatics, Institute of Biomedical Chemistry, 107076 Moscow, Russia; (S.I.); (D.A.F.); (V.P.)
| |
Collapse
|
778
|
Russell SM, Alba-Patiño A, Barón E, Borges M, Gonzalez-Freire M, de la Rica R. Biosensors for Managing the COVID-19 Cytokine Storm: Challenges Ahead. ACS Sens 2020; 5:1506-1513. [PMID: 32482077 PMCID: PMC7299396 DOI: 10.1021/acssensors.0c00979] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022]
Abstract
The global COVID-19 pandemic has oversaturated many intensive care units to the point of collapse, leading to enormous spikes in death counts. Before critical care becomes a necessity, identifying patients who are likely to become critically ill and providing prompt treatment is a strategy to avoid ICU oversaturation. There is a consensus that a hyperinflammatory syndrome or a "cytokine storm" is responsible for poor outcomes in COVID-19. Measuring cytokine levels at the point of care is required in order to better understand this process. In this Perspective, we summarize the main events behind the cytokine storm in COVID-19 as well as current experimental treatments. We advocate for a new biosensor-enabled paradigm to personalize the management of COVID-19 and stratify patients. Biosensor-guided dosing and timing of immunomodulatory therapies could maximize the benefits of these anti-inflammatory treatments while minimizing deleterious effects. Biosensors will also be essential in order to detect complications such as coinfections and sepsis, which are common in immunosuppressed patients. Finally, we propose the ideal features of these biosensors using some prototypes from the recent literature as examples. Multisensors, lateral flow tests, mobile biosensors, and wearable biosensors are seen as key players for precision medicine in COVID-19.
Collapse
Affiliation(s)
- Steven M. Russell
- Multidisciplinary Sepsis Group and
Vascular and Metabolic Pathologies Group,
Health Research Institute of the Balearic Islands
(IdISBa), Son Espases University Hospital, S
building, Ctra. de Valldemossa 79, 07120 Palma de Mallorca,
Spain
| | - Alejandra Alba-Patiño
- Multidisciplinary Sepsis Group and
Vascular and Metabolic Pathologies Group,
Health Research Institute of the Balearic Islands
(IdISBa), Son Espases University Hospital, S
building, Ctra. de Valldemossa 79, 07120 Palma de Mallorca,
Spain
| | - Enrique Barón
- Multidisciplinary Sepsis Group and
Vascular and Metabolic Pathologies Group,
Health Research Institute of the Balearic Islands
(IdISBa), Son Espases University Hospital, S
building, Ctra. de Valldemossa 79, 07120 Palma de Mallorca,
Spain
| | - Marcio Borges
- Multidisciplinary Sepsis Group and
Vascular and Metabolic Pathologies Group,
Health Research Institute of the Balearic Islands
(IdISBa), Son Espases University Hospital, S
building, Ctra. de Valldemossa 79, 07120 Palma de Mallorca,
Spain
- Multidisciplinary Sepsis Unit, ICU,
Son Llàtzer University
Hospital, 07198 Palma de Mallorca,
Spain
| | - Marta Gonzalez-Freire
- Multidisciplinary Sepsis Group and
Vascular and Metabolic Pathologies Group,
Health Research Institute of the Balearic Islands
(IdISBa), Son Espases University Hospital, S
building, Ctra. de Valldemossa 79, 07120 Palma de Mallorca,
Spain
| | - Roberto de la Rica
- Multidisciplinary Sepsis Group and
Vascular and Metabolic Pathologies Group,
Health Research Institute of the Balearic Islands
(IdISBa), Son Espases University Hospital, S
building, Ctra. de Valldemossa 79, 07120 Palma de Mallorca,
Spain
| |
Collapse
|
779
|
Nasonov EL, Lila AM. BARICITINIB: NEW PHARMACOTHERAPY OPTIONS FOR RHEUMATOID ARTHRITIS AND OTHER IMMUNE-MEDIATED INFLAMMATORY RHEUMATIC DISEASES. RHEUMATOLOGY SCIENCE AND PRACTICE 2020. [DOI: 10.14412/1995-4484-2020-304-316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Deciphering the mechanisms of the pathogenesis of immune-mediated inflammatory rheumatic diseases (IMIRDs) in conjunction with designing a wide range of biological agents is one of the major medical advances in the 21st century. A new promising area of pharmacotherapy for IMIRDs is associated with the design of the so-called targeted oral medications that primarily include Janus kinase (JAK) inhibitors. The review presents new data on the efficacy and safety of the new JAK inhibitor baricitinib in treating rheumatoid arthritis and other IMIRDs.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology;
I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| | - A. M. Lila
- V.A. Nasonova Research Institute of Rheumatology; Russian Medical Academy of
Continuing Professional Education, Ministry of Health of Russia
| |
Collapse
|
780
|
Nasonov EL. IMMUNOPATHOLOGY AND IMMUNOPHARMACOTHERAPY OF CORONAVIRUS DISEASE 2019 (COVID-19): FOCUS ON INTERLEUKIN 6. RHEUMATOLOGY SCIENCE AND PRACTICE 2020. [DOI: 10.14412/1995-4484-2020-245-261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic has drawn closer attention than ever before to the problems of the immunopathology of human diseases, many of which have been reflected when studying immune-mediated inflammatory rheumatic diseases (IIRDs). The hyperimmune response called a cytokine storm, the pathogenetic subtypes of which include hemophagocytic lymphohistiocytosis, macrophage activation syndrome, and cytokine release syndrome, is among the most serious complications of IIRDs or treatment for malignant neoplasms and may be a stage of COVID-19 progression. A premium is placed to interleukin-6 (IL-6) in the spectrum of cytokines involved in the pathogenesis of the cytokine storm syndrome. The clinical introduction of monoclonal antibodies (mAbs) that inhibit the activity of this cytokine (tocilizumab, sarilumab, etc.) is one of the major advances in the treatment of IIRDs and critical conditions within the cytokine storm syndrome in COVID-19. The review discusses data on the clinical and prognostic value of IL-6 and the effectiveness of anti-IL-6 receptor and anti-IL-6 mAbs, as well as prospects for personalized therapy of the cytokine storm syndrome in COVID-19.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology; I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| |
Collapse
|
781
|
Guaraldi G, Meschiari M, Cozzi-Lepri A, Milic J, Tonelli R, Menozzi M, Franceschini E, Cuomo G, Orlando G, Borghi V, Santoro A, Di Gaetano M, Puzzolante C, Carli F, Bedini A, Corradi L, Fantini R, Castaniere I, Tabbì L, Girardis M, Tedeschi S, Giannella M, Bartoletti M, Pascale R, Dolci G, Brugioni L, Pietrangelo A, Cossarizza A, Pea F, Clini E, Salvarani C, Massari M, Viale PL, Mussini C. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. LANCET RHEUMATOLOGY 2020; 2:e474-e484. [PMID: 32835257 PMCID: PMC7314456 DOI: 10.1016/s2665-9913(20)30173-9] [Citation(s) in RCA: 682] [Impact Index Per Article: 136.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background No therapy is approved for COVID-19 pneumonia. The aim of this study was to assess the role of tocilizumab in reducing the risk of invasive mechanical ventilation and death in patients with severe COVID-19 pneumonia who received standard of care treatment. Methods This retrospective, observational cohort study included adults (≥18 years) with severe COVID-19 pneumonia who were admitted to tertiary care centres in Bologna and Reggio Emilia, Italy, between Feb 21 and March 24, 2020, and a tertiary care centre in Modena, Italy, between Feb 21 and April 30, 2020. All patients were treated with the standard of care (ie, supplemental oxygen, hydroxychloroquine, azithromycin, antiretrovirals, and low molecular weight heparin), and a non-randomly selected subset of patients also received tocilizumab. Tocilizumab was given either intravenously at 8 mg/kg bodyweight (up to a maximum of 800 mg) in two infusions, 12 h apart, or subcutaneously at 162 mg administered in two simultaneous doses, one in each thigh (ie, 324 mg in total), when the intravenous formulation was unavailable. The primary endpoint was a composite of invasive mechanical ventilation or death. Treatment groups were compared using Kaplan-Meier curves and Cox regression analysis after adjusting for sex, age, recruiting centre, duration of symptoms, and baseline Sequential Organ Failure Assessment (SOFA) score. Findings Of 1351 patients admitted, 544 (40%) had severe COVID-19 pneumonia and were included in the study. 57 (16%) of 365 patients in the standard care group needed mechanical ventilation, compared with 33 (18%) of 179 patients treated with tocilizumab (p=0·41; 16 [18%] of 88 patients treated intravenously and 17 [19%] of 91 patients treated subcutaneously). 73 (20%) patients in the standard care group died, compared with 13 (7%; p<0·0001) patients treated with tocilizumab (six [7%] treated intravenously and seven [8%] treated subcutaneously). After adjustment for sex, age, recruiting centre, duration of symptoms, and SOFA score, tocilizumab treatment was associated with a reduced risk of invasive mechanical ventilation or death (adjusted hazard ratio 0·61, 95% CI 0·40–0·92; p=0·020). 24 (13%) of 179 patients treated with tocilizumab were diagnosed with new infections, versus 14 (4%) of 365 patients treated with standard of care alone (p<0·0001). Interpretation Treatment with tocilizumab, whether administered intravenously or subcutaneously, might reduce the risk of invasive mechanical ventilation or death in patients with severe COVID-19 pneumonia. Funding None.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marianna Meschiari
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Alessandro Cozzi-Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, University College London, London, UK
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Tonelli
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Marianna Menozzi
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Erica Franceschini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Gianluca Cuomo
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Gabriella Orlando
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Vanni Borghi
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Antonella Santoro
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Margherita Di Gaetano
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Cinzia Puzzolante
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Federica Carli
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Andrea Bedini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Luca Corradi
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Riccardo Fantini
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Ivana Castaniere
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Tabbì
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Massimo Girardis
- Department of Anaesthesia and Intensive Care Unit, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara Tedeschi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maddalena Giannella
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Michele Bartoletti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Renato Pascale
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Dolci
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Lucio Brugioni
- Internal Medicine Department, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy
| | - Antonello Pietrangelo
- Internal Medicine Department, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Pea
- Institute of Clinical Pharmacology, Azienda Ospedaliero-Universitaria Santa Maria Della Misericordia, University of Udine, Udine, Italy
| | - Enrico Clini
- Respiratory Diseases Unit, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Salvarani
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Department of Rheumatology, Azienda USL-IRCCS di Reggio Emilia, Reggio nell'Emilia, Italy
| | - Marco Massari
- Infectious Disease Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio nell'Emilia, Italy
| | - Pier Luigi Viale
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Cristina Mussini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena, Italy.,Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
782
|
Tsuchiya A, Takeuchi S, Iwasawa T, Kumagai M, Sato T, Motegi S, Ishii Y, Koseki Y, Tomiyoshi K, Natsui K, Takeda N, Yoshida Y, Yamazaki F, Kojima Y, Watanabe Y, Kimura N, Tominaga K, Kamimura H, Takamura M, Terai S. Therapeutic potential of mesenchymal stem cells and their exosomes in severe novel coronavirus disease 2019 (COVID-19) cases. Inflamm Regen 2020; 40:14. [PMID: 32582401 PMCID: PMC7306412 DOI: 10.1186/s41232-020-00121-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
The novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19) and the ensuing worldwide pandemic. The spread of the virus has had global effects such as activity restriction, economic stagnation, and collapse of healthcare infrastructure. Severe SARS-CoV-2 infection induces a cytokine storm, leading to acute respiratory distress syndrome (ARDS) and multiple organ failure, which are very serious health conditions and must be mitigated or resolved as soon as possible. Mesenchymal stem cells (MSCs) and their exosomes can affect immune cells by inducing anti-inflammatory macrophages, regulatory T and B cells, and regulatory dendritic cells, and can inactivate T cells. Hence, they are potential candidate agents for treatment of severe cases of COVID-19. In this review, we report the background of severe cases of COVID-19, basic aspects and mechanisms of action of MSCs and their exosomes, and discuss basic and clinical studies based on MSCs and exosomes for influenza-induced ARDS. Finally, we report the potential of MSC and exosome therapy in severe cases of COVID-19 in recently initiated or planned clinical trials of MSCs (33 trials) and exosomes (1 trial) registered in 13 countries on ClinicalTrials.gov.
Collapse
Affiliation(s)
- Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Suguru Takeuchi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Takahiro Iwasawa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Masaru Kumagai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Takeki Sato
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Satoko Motegi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Yui Ishii
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Youhei Koseki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Kei Tomiyoshi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Kazuki Natsui
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Nobutaka Takeda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Yuki Yoshida
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Fusako Yamazaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Yuichi Kojima
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Yusuke Watanabe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Naruhiro Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Kentaro Tominaga
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Masaaki Takamura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8510 Japan
| |
Collapse
|
783
|
Smetana K, Brábek J. Role of Interleukin-6 in Lung Complications in Patients With COVID-19: Therapeutic Implications. In Vivo 2020; 34:1589-1592. [PMID: 32503815 DOI: 10.21873/invivo.11947] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 12/22/2022]
Abstract
COVID-19 is viral respiratory infection with frequently fatal lung complications in the elderly or in people with serious comorbidities. Lung destruction appears to be associated with a cytokine storm related to an increased level of interleukin-6 (IL6). Therapeutic targeting of the interleukin-6 signaling pathway can attenuate such a cytokine storm and can be beneficial for patients with COVID-19 in danger of pulmonary failure. This article demonstrates the importance of IL6 in progression of disease and the possibility of inhibition of IL6 signaling in COVID-19 therapy.
Collapse
Affiliation(s)
- Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic .,BIOCEV, Vestec, Czech Republic
| | - Jan Brábek
- BIOCEV, Vestec, Czech Republic .,Department of Cell Biology, Faculty of Sciences, Charles University, Prague, Czech Republic
| |
Collapse
|
784
|
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease, caused by severe acute respiratory syndrome coronavirus 2, which predominantly affects the lungs and, under certain circumstances, leads to an excessive or uncontrolled immune activation and cytokine response in alveolar structures. The pattern of pro-inflammatory cytokines induced in COVID-19 has similarities to those targeted in the treatment of rheumatoid arthritis. Several clinical studies are underway that test the effects of inhibiting IL-6, IL-1β or TNF or targeting cytokine signalling via Janus kinase inhibition in the treatment of COVID-19. Despite these similarities, COVID-19 and other zoonotic coronavirus-mediated diseases do not induce clinical arthritis, suggesting that a local inflammatory niche develops in alveolar structures and drives the disease process. COVID-19 constitutes a challenge for patients with inflammatory arthritis for several reasons, in particular, the safety of immune interventions during the pandemic. Preliminary data, however, do not suggest that patients with inflammatory arthritis are at increased risk of COVID-19. This Perspective article explores similarities in the inflammatory processes underlying coronavirus disease 2019 (COVID-19) and rheumatoid arthritis, including the role of pro-inflammatory cytokines and the potential of anti-cytokine therapies to treat COVID-19, as well as the effect of the COVID-19 pandemic on rheumatology.
Collapse
|
785
|
Belladonna ML, Orabona C. Potential Benefits of Tryptophan Metabolism to the Efficacy of Tocilizumab in COVID-19. Front Pharmacol 2020; 11:959. [PMID: 32636755 PMCID: PMC7319082 DOI: 10.3389/fphar.2020.00959] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/11/2020] [Indexed: 12/30/2022] Open
Abstract
Tocilizumab has been proposed as a means of opposing hyperinflammatory responses in intensive care patients with COVID-19. Here, we briefly discuss the potentially multiple, synergistic mechanisms whereby tocilizumab might exert therapeutic activity, mostly focusing on the production of tryptophan-derived catabolites that would result from blockade of IL-6 signaling, as contextualized to the cytokine storm occurring in COVID-19 patients.
Collapse
Affiliation(s)
| | - Ciriana Orabona
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
786
|
Sano M, Ichihara G, Katsumata Y, Hiraide T, Hirai A, Momoi M, Tamura T, Ohata S, Kobayashi E. Pharmacokinetics of a single inhalation of hydrogen gas in pigs. PLoS One 2020; 15:e0234626. [PMID: 32559239 PMCID: PMC7304914 DOI: 10.1371/journal.pone.0234626] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
The benefits of inhaling hydrogen gas (H2) have been widely reported but its pharmacokinetics have not yet been sufficiently analyzed. We developed a new experimental system in pigs to closely evaluate the process by which H2 is absorbed in the lungs, enters the bloodstream, and is distributed, metabolized, and excreted. We inserted and secured catheters into the carotid artery (CA), portal vein (PV), and supra-hepatic inferior vena cava (IVC) to allow repeated blood sampling and performed bilateral thoracotomy to collapse the lungs. Then, using a hydrogen-absorbing alloy canister, we filled the lungs to the maximum inspiratory level with 100% H2. The pig was maintained for 30 seconds without resuming breathing, as if they were holding their breath. We collected blood from the three intravascular catheters after 0, 3, 10, 30, and 60 minutes and measured H2 concentration by gas chromatography. H2 concentration in the CA peaked immediately after breath holding; 3 min later, it dropped to 1/40 of the peak value. Peak H2 concentrations in the PV and IVC were 40% and 14% of that in the CA, respectively. However, H2 concentration decay in the PV and IVC (half-life: 310 s and 350 s, respectively) was slower than in the CA (half-life: 92 s). At 10 min, H2 concentration was significantly higher in venous blood than in arterial blood. At 60 min, H2 was detected in the portal blood at a concentration of 6.9-53 nL/mL higher than at steady state, and in the SVC 14-29 nL/mL higher than at steady state. In contrast, H2 concentration in the CA decreased to steady state levels. This is the first report showing that inhaled H2 is transported to the whole body by advection diffusion and metabolized dynamically.
Collapse
Affiliation(s)
- Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Center for Molecular Hydrogen Medicine, Keio University, Minato-ku, Tokyo, Japan
| | - Genki Ichihara
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Center for Molecular Hydrogen Medicine, Keio University, Minato-ku, Tokyo, Japan
| | - Yoshinori Katsumata
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Center for Molecular Hydrogen Medicine, Keio University, Minato-ku, Tokyo, Japan
| | - Takahiro Hiraide
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Akeo Hirai
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Mizuki Momoi
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tomoyoshi Tamura
- Center for Molecular Hydrogen Medicine, Keio University, Minato-ku, Tokyo, Japan
- Department of Emergency and Critical Care Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Shigeo Ohata
- Department of Neurology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Eiji Kobayashi
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Center for Molecular Hydrogen Medicine, Keio University, Minato-ku, Tokyo, Japan
- Department of Organ Fabrication, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
787
|
Abstract
Purpose of Review An unprecedented outbreak of the novel coronavirus in China (COVID-19) occurred in December 2019, and then engulfed the entire world, presenting a significant and urgent threat to global health. Many research institutes have been involved in the development of drugs and vaccines against COVID-19. Recent Findings At present, the strategy of new use of old drugs is mainly used to screen candidate drugs against the novel coronavirus (later termed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) and inhibit excessive immune response. Related research has made great progress. Summary In this review, we summarize the drugs used for COVID-19 treatment in China based on the emerging basic and clinical data. It is hoped that this review will be useful to provide guidance for the prevention, treatment, and control of COVID-19.
Collapse
Affiliation(s)
- Linzi Fan
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang People's Republic of China
| | - Shuang Jiang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang People's Republic of China
| | - Xinrong Yang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang People's Republic of China
| | - Zhibin Wang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin, 150040 Heilongjiang China
| | - Chunjuan Yang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, 150081 Heilongjiang People's Republic of China
| |
Collapse
|
788
|
McAuliffe S, Ray S, Fallon E, Bradfield J, Eden T, Kohlmeier M. Dietary micronutrients in the wake of COVID-19: an appraisal of evidence with a focus on high-risk groups and preventative healthcare. BMJ Nutr Prev Health 2020; 3:93-99. [PMID: 33235973 PMCID: PMC7664499 DOI: 10.1136/bmjnph-2020-000100] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/23/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
Existing micronutrient deficiencies, even if only a single micronutrient, can impair immune function and increase susceptibility to infectious disease. Certain population groups are more likely to have micronutrient deficiencies, while certain disease pathologies and treatment practices also exacerbate risk, meaning these groups tend to suffer increased morbidity and mortality from infectious diseases. Optimisation of overall nutritional status, including micronutrients, can be effective in reducing incidence of infectious disease. Micronutrient deficiencies are rarely recognised but are prevalent in the UK, as well as much more widely, particularly in high-risk groups susceptible to COVID-19. Practitioners should be aware of this fact and should make it a consideration for the screening process in COVID-19, or when screening may be difficult or impractical, to ensure blanket treatment as per the best practice guidelines. Correction of established micronutrient deficiencies, or in some cases assumed suboptimal status, has the potential to help support immune function and mitigate risk of infection. The effects of and immune response to COVID-19 share common characteristics with more well-characterised severe acute respiratory infections. Correction of micronutrient deficiencies has proven effective in several infectious diseases and has been shown to promote favourable clinical outcomes. Micronutrients appear to play key roles in mediating the inflammatory response and such effects may be enhanced through correction of deficiencies. Many of those at highest risk during the COVID-19 pandemic are also populations at highest risk of micronutrient deficiencies and poorer overall nutrition. Correction of micronutrient deficiencies in established COVID-19 infection may contribute to supporting immune response to infection in those at highest risk. There is a need for further research to establish optimal public health practice and clinical intervention regimens.
Collapse
Affiliation(s)
- Shane McAuliffe
- NNEdPro Global Centre for Nutrition and Health, St John’s Innovation Centre, Cambridge, UK
| | - Sumantra Ray
- NNEdPro Global Centre for Nutrition and Health, St John’s Innovation Centre, Cambridge, UK
- School of Biomedical Sciences, Ulster University at Coleraine, Coleraine, Northern Ireland
- School of Humanities and Social Sciences, University of Cambridge, Cambridge, UK
| | - Emily Fallon
- NNEdPro Global Centre for Nutrition and Health, St John’s Innovation Centre, Cambridge, UK
- Maldon District Council, Council Offices, Maldon, UK
| | - James Bradfield
- NNEdPro Global Centre for Nutrition and Health, St John’s Innovation Centre, Cambridge, UK
| | - Timothy Eden
- NNEdPro Global Centre for Nutrition and Health, St John’s Innovation Centre, Cambridge, UK
- Imperial College London NHS Foundation Trust, London, UK
| | - Martin Kohlmeier
- NNEdPro Global Centre for Nutrition and Health, St John’s Innovation Centre, Cambridge, UK
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| |
Collapse
|
789
|
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic infection caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). COVID-19 significantly affects multiple systems including the cardiovascular system. Most importantly, in addition to the direct injury from the virus per se, the subsequent cytokine storm, an overproduction of immune cells and their activating compounds, causes devastating damage. To date, emerging anti-SARS-CoV-2 treatments are warranted to control epidemics. Several candidate drugs have been screened and are currently under investigation. These primarily include antiviral regimens and immunomodulatory regimens. However, beyond the anti-SARS-CoV-2 effects, these drugs may also have risks to the cardiovascular system, especially altering cardiac conduction. Herein, we review the cardiovascular risks of potential anti-COVID-19 drugs.
Collapse
|
790
|
Villena J, Kitazawa H. The Modulation of Mucosal Antiviral Immunity by Immunobiotics: Could They Offer Any Benefit in the SARS-CoV-2 Pandemic? Front Physiol 2020; 11:699. [PMID: 32670091 PMCID: PMC7326040 DOI: 10.3389/fphys.2020.00699] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023] Open
Abstract
Viral respiratory infections are of major importance because of their capacity to cause of a high degree of morbidity and mortality in high-risk populations, and to rapidly spread between countries. Perhaps the best example of this global threat is the infectious disease caused by the new SARS-CoV-2 virus, which has infected more than 4 million people worldwide, causing the death of 287,000 persons according to the WHO's situation report on May 13, 2020. The availability of therapeutic tools that would be used massively to prevent or mitigate the detrimental effects of emerging respiratory viruses on human health is therefore mandatory. In this regard, research from the last decade has reported the impact of the intestinal microbiota on the respiratory immunity. It was conclusively demonstrated how the variations in the intestinal microbiota affect the responses of respiratory epithelial cells and antigen presenting cells against respiratory virus attack. Moreover, the selection of specific microbial strains (immunobiotics) with the ability to modulate immunity in distal mucosal sites made possible the generation of nutritional interventions to strengthen respiratory antiviral defenses. In this article, the most important characteristics of the limited information available regarding the immune response against SARS-CoV-2 virus are revised briefly. In addition, this review summarizes the knowledge on the cellular and molecular mechanisms involved in the improvement of respiratory antiviral defenses by beneficial immunobiotic microorganisms such as Lactobacillus rhamnosus CRL1505. The ability of beneficial microorganisms to enhance type I interferons and antiviral factors in the respiratory tract, stimulate Th1 response and antibodies production, and regulate inflammation and coagulation activation during the course of viral infections reducing tissue damage and preserving lung functionally, clearly indicate the potential of immunobiotics to favorably influence the immune response against SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán, Argentina
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
791
|
García LF. Immune Response, Inflammation, and the Clinical Spectrum of COVID-19. Front Immunol 2020; 11:1441. [PMID: 32612615 PMCID: PMC7308593 DOI: 10.3389/fimmu.2020.01441] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/03/2020] [Indexed: 01/08/2023] Open
Abstract
The current COVID-19 pandemic began in December 2019 in Wuhan (China) and rapidly extended to become a global sanitary and economic emergency. Its etiological agent is the coronavirus SARS-CoV-2. COVID-19 presents a wide spectrum of clinical manifestations, which ranges from an asymptomatic infection to a severe pneumonia accompanied by multisystemic failure that can lead to a patient's death. The immune response to SARS-CoV-2 is known to involve all the components of the immune system that together appear responsible for viral elimination and recovery from the infection. Nonetheless, such immune responses are implicated in the disease's progression to a more severe and lethal process. This review describes the general aspects of both COVID-19 and its etiological agent SARS-CoV-2, stressing the similarities with other severe coronavirus infections, such as SARS and MERS, but more importantly, pointing toward the evidence supporting the hypothesis that the clinical spectrum of COVID-19 is a consequence of the corresponding variable spectrum of the immune responses to the virus. The critical point where progression of the disease ensues appears to center on loss of the immune regulation between protective and altered responses due to exacerbation of the inflammatory components. Finally, it appears possible to delineate certain major challenges deserving of exhaustive investigation to further understand COVID-19 immunopathogenesis, thus helping to design more effective diagnostic, therapeutic, and prophylactic strategies.
Collapse
Affiliation(s)
- Luis F. García
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
792
|
McCord JM, Hybertson BM, Cota-Gomez A, Geraci KP, Gao B. Nrf2 Activator PB125 ® as a Potential Therapeutic Agent against COVID-19. Antioxidants (Basel) 2020; 9:E518. [PMID: 32545518 PMCID: PMC7346195 DOI: 10.3390/antiox9060518] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Nrf2 is a transcription factor that regulates cellular redox balance and the expression of a wide array of genes involved in immunity and inflammation, including antiviral actions. Nrf2 activity declines with age, making the elderly more susceptible to oxidative stress-mediated diseases, which include type 2 diabetes, chronic inflammation, and viral infections. Published evidence suggests that Nrf2 activity may regulate important mechanisms affecting viral susceptibility and replication. We examined gene expression levels by GeneChip microarray and by RNA-seq assays. We found that the potent Nrf2-activating composition PB125® downregulates ACE2 and TMPRSS2 mRNA expression in human liver-derived HepG2 cells. ACE2 is a surface receptor and TMPRSS2 activates the spike protein for SARS-CoV-2 entry into host cells. Furthermore, in endotoxin-stimulated primary human pulmonary artery endothelial cells, we report the marked downregulation by PB125 of 36 genes encoding cytokines. These include IL-1-beta, IL-6, TNF-α, the cell adhesion molecules ICAM-1, VCAM-1, and E-selectin, and a group of IFN-γ-induced genes. Many of these cytokines have been specifically identified in the "cytokine storm" observed in fatal cases of COVID-19, suggesting that Nrf2 activation may significantly decrease the intensity of the storm.
Collapse
Affiliation(s)
- Joe M. McCord
- Pathways Bioscience, Aurora, CO 80045, USA; (B.M.H.); (B.G.)
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.C.-G.); (K.P.G.)
| | - Brooks M. Hybertson
- Pathways Bioscience, Aurora, CO 80045, USA; (B.M.H.); (B.G.)
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.C.-G.); (K.P.G.)
| | - Adela Cota-Gomez
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.C.-G.); (K.P.G.)
| | - Kara P. Geraci
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.C.-G.); (K.P.G.)
| | - Bifeng Gao
- Pathways Bioscience, Aurora, CO 80045, USA; (B.M.H.); (B.G.)
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.C.-G.); (K.P.G.)
| |
Collapse
|
793
|
McCord JM, Hybertson BM, Cota-Gomez A, Gao B. Nrf2 Activator PB125® as a Potential Therapeutic Agent Against COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511372 PMCID: PMC7263501 DOI: 10.1101/2020.05.16.099788] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nrf2 is a transcription factor that regulates cellular redox balance and the expression of a wide array of genes involved in immunity and inflammation, including antiviral actions. Nrf2 activity declines with age, making the elderly more susceptible to oxidative stress-mediated diseases, which include type 2 diabetes, chronic inflammation, and viral infections. Published evidence suggests that Nrf2 activity may regulate important mechanisms affecting viral susceptibility and replication. We examined gene expression levels by GeneChip microarray and by RNA-seq assays. We found that the potent Nrf2 activating composition PB125® downregulates ACE2 and TMPRSS2 mRNA expression in human liver-derived HepG2 cells. ACE2 is a surface receptor and TMPRSS2 activates the spike protein for SARS-Cov-2 entry into host cells. Furthermore, in endotoxin-stimulated primary human pulmonary artery endothelial cells we report the marked downregulation by PB125 of 36 genes encoding cytokines. These include IL1-beta, IL6, TNF-α the cell adhesion molecules ICAM1, VCAM1, and E-selectin, and a group of IFN-γ-induced genes. Many of these cytokines have been specifically identified in the “cytokine storm” observed in fatal cases of COVID-19, suggesting that Nrf2 activation may significantly decrease the intensity of the storm.
Collapse
Affiliation(s)
- Joe M McCord
- Pathways Bioscience, Aurora, CO 80045, USA.,Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brooks M Hybertson
- Pathways Bioscience, Aurora, CO 80045, USA.,Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Adela Cota-Gomez
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bifeng Gao
- Pathways Bioscience, Aurora, CO 80045, USA.,Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
794
|
Al-Qudsi O, Whitson B, Bhatt A, Chucta S, Tripathi R. A case of cytokine hemofiltration and extracorporeal life support as treatment of coronavirus disease 2019 (COVID-19) respiratory failure. J Thorac Cardiovasc Surg 2020; 161:S0022-5223(20)31318-0. [PMID: 32624302 PMCID: PMC7282777 DOI: 10.1016/j.jtcvs.2020.05.082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Omar Al-Qudsi
- Section of Critical Care Medicine, Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| | - Bryan Whitson
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Amar Bhatt
- Section of Critical Care Medicine, Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Sheila Chucta
- Department of Nursing, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ravi Tripathi
- Section of Critical Care Medicine, Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
795
|
He LH, Ren LF, Li JF, Wu YN, Li X, Zhang L. Intestinal Flora as a Potential Strategy to Fight SARS-CoV-2 Infection. Front Microbiol 2020; 11:1388. [PMID: 32582138 PMCID: PMC7295895 DOI: 10.3389/fmicb.2020.01388] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/29/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread rapidly worldwide, seriously endangering human health. In addition to the typical symptoms of pulmonary infection, patients with COVID-19 have been reported to have gastrointestinal symptoms and/or intestinal flora dysbiosis. It is known that a healthy intestinal flora is closely related to the maintenance of pulmonary and systemic health by regulating the host immune homeostasis. Role of the "gut-lung axis" has also been well-articulated. This review provides a novel suggestion that intestinal flora may be one of the mediators of the gastrointestinal responses and abnormal immune responses in hosts caused by SARS-CoV-2; improving the composition of intestinal flora and the proportion of its metabolites through probiotics, and personalized diet could be a potential strategy to prevent and treat COVID-19. More clinical and evidence-based medical trials may be initiated to determine the strategy.
Collapse
Affiliation(s)
- Li-Hong He
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation Gansu Province, Lanzhou, China
| | - Long-Fei Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation Gansu Province, Lanzhou, China
| | - Jun-Feng Li
- The Department of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yong-Na Wu
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation Gansu Province, Lanzhou, China
| | - Xun Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation Gansu Province, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation Gansu Province, Lanzhou, China
| |
Collapse
|
796
|
Smith JC, Sausville EL, Girish V, Yuan ML, Vasudevan A, John KM, Sheltzer JM. Cigarette Smoke Exposure and Inflammatory Signaling Increase the Expression of the SARS-CoV-2 Receptor ACE2 in the Respiratory Tract. Dev Cell 2020; 53:514-529.e3. [PMID: 32425701 PMCID: PMC7229915 DOI: 10.1016/j.devcel.2020.05.012] [Citation(s) in RCA: 282] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 01/06/2023]
Abstract
The factors mediating fatal SARS-CoV-2 infections are poorly understood. Here, we show that cigarette smoke causes a dose-dependent upregulation of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 receptor, in rodent and human lungs. Using single-cell sequencing data, we demonstrate that ACE2 is expressed in a subset of secretory cells in the respiratory tract. Chronic smoke exposure triggers the expansion of this cell population and a concomitant increase in ACE2 expression. In contrast, quitting smoking decreases the abundance of these secretory cells and reduces ACE2 levels. Finally, we demonstrate that ACE2 expression is responsive to inflammatory signaling and can be upregulated by viral infections or interferon treatment. Taken together, these results may partially explain why smokers are particularly susceptible to severe SARS-CoV-2 infections. Furthermore, our work identifies ACE2 as an interferon-stimulated gene in lung cells, suggesting that SARS-CoV-2 infections could create positive feedback loops that increase ACE2 levels and facilitate viral dissemination.
Collapse
Affiliation(s)
- Joan C Smith
- Google, Inc., New York City, NY 10011, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Erin L Sausville
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Vishruth Girish
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Stony Brook University, Stony Brook, NY 11794, USA
| | - Monet Lou Yuan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anand Vasudevan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kristen M John
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Hofstra University, Hempstead, NY 11549, USA
| | - Jason M Sheltzer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
797
|
Silibinin and SARS-CoV-2: Dual Targeting of Host Cytokine Storm and Virus Replication Machinery for Clinical Management of COVID-19 Patients. J Clin Med 2020; 9:jcm9061770. [PMID: 32517353 PMCID: PMC7356916 DOI: 10.3390/jcm9061770] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/05/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19, the illness caused by infection with the novel coronavirus SARS-CoV-2, is a rapidly spreading global pandemic in urgent need of effective treatments. Here we present a comprehensive examination of the host- and virus-targeted functions of the flavonolignan silibinin, a potential drug candidate against COVID-19/SARS-CoV-2. As a direct inhibitor of STAT3—a master checkpoint regulator of inflammatory cytokine signaling and immune response—silibinin might be expected to phenotypically integrate the mechanisms of action of IL-6-targeted monoclonal antibodies and pan-JAK1/2 inhibitors to limit the cytokine storm and T-cell lymphopenia in the clinical setting of severe COVID-19. As a computationally predicted, remdesivir-like inhibitor of RNA-dependent RNA polymerase (RdRp)—the central component of the replication/transcription machinery of SARS-CoV-2—silibinin is expected to reduce viral load and impede delayed interferon responses. The dual ability of silibinin to target both the host cytokine storm and the virus replication machinery provides a strong rationale for the clinical testing of silibinin against the COVID-19 global public health emergency. A randomized, open-label, phase II multicentric clinical trial (SIL-COVID19) will evaluate the therapeutic efficacy of silibinin in the prevention of acute respiratory distress syndrome in moderate-to-severe COVID-19-positive onco-hematological patients at the Catalan Institute of Oncology in Catalonia, Spain.
Collapse
|
798
|
Narasaraju T, Tang BM, Herrmann M, Muller S, Chow VTK, Radic M. Neutrophilia and NETopathy as Key Pathologic Drivers of Progressive Lung Impairment in Patients With COVID-19. Front Pharmacol 2020; 11:870. [PMID: 32581816 PMCID: PMC7291833 DOI: 10.3389/fphar.2020.00870] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022] Open
Abstract
There is an urgent need for new therapeutic strategies to contain the spread of the novel coronavirus disease 2019 (COVID-19) and to curtail its most severe complications. Severely ill patients experience pathologic manifestations of acute respiratory distress syndrome (ARDS), and clinical reports demonstrate striking neutrophilia, elevated levels of multiple cytokines, and an exaggerated inflammatory response in fatal COVID-19. Mechanical respirator devices are the most widely applied therapy for ARDS in COVID-19, yet mechanical ventilation achieves strikingly poor survival. Many patients, who recover, experience impaired cognition or physical disability. In this review, we argue the need to develop therapies aimed at inhibiting neutrophil recruitment, activation, degranulation, and neutrophil extracellular trap (NET) release. Moreover, we suggest that currently available pharmacologic approaches should be tested as treatments for ARDS in COVID-19. In our view, targeting host-mediated immunopathology holds promise to alleviate progressive pathologic complications of ARDS and reduce morbidities and mortalities in severely ill patients with COVID-19.
Collapse
Affiliation(s)
- Teluguakula Narasaraju
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, United States
| | - Benjamin M. Tang
- Department of Intensive Care Medicine, Nepean Hospital, Sydney, NSW, Australia
| | - Martin Herrmann
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France
- Laboratory of Excellence Medalis, Institut de science et d'ingénierie supramoléculaire, and University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| | - Vincent T. K. Chow
- Department of Microbiology and Immunology, Infectious Diseases Program, School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
799
|
|
800
|
Olivari L, Riccardi N, Rodari P, Angheben A, Artioli P, Salgarello M. COVID-19 pneumonia: increased choline uptake with 18F-choline PET/CT. Eur J Nucl Med Mol Imaging 2020; 47:2476-2477. [PMID: 32500168 PMCID: PMC7272208 DOI: 10.1007/s00259-020-04870-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Laura Olivari
- Department of Nuclear Medicine, IRCCS Ospedale Sacro Cuore Don Calabria, Via Don Angelo Sempreboni, 5- Negrar di Valpolicella, Verona, Italy.
| | - Niccolò Riccardi
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Paola Rodari
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Andrea Angheben
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Paolo Artioli
- Nuclear Medicine, University of Padova, Padova, Italy
| | - Matteo Salgarello
- Department of Nuclear Medicine, IRCCS Ospedale Sacro Cuore Don Calabria, Via Don Angelo Sempreboni, 5- Negrar di Valpolicella, Verona, Italy
| |
Collapse
|